Back to Search Start Over

Mammalian enabled protein enhances tamoxifen sensitivity of the hormone receptor-positive breast cancer patients by suppressing the AKT signaling pathway.

Authors :
He, Lifang
She, Chuanghong
Jiang, Sen
Qi, Zhaochang
Deng, Zihao
Ji, Likeng
Cui, Yukun
Wu, Jundong
Source :
Biology Direct; 3/8/2024, Vol. 19 Issue 1, p1-14, 14p
Publication Year :
2024

Abstract

Background: Mammalian enabled (MENA) protein is a member of the enabled/vasodilator stimulated phosphoprotein (Ena/VASP) protein family, which regulates cytoplasmic actin network assembly. It plays a significant role in breast cancer invasion, migration, and resistance against targeted therapy and chemotherapy. However, its role in the efficacy of endocrine therapy for the hormone receptor-positive (HR<superscript>+</superscript>) breast cancer patients is not known. This study investigated the role of MENA in the resistance against tamoxifen therapy in patients with HR<superscript>+</superscript> breast cancer and the underlying mechanisms. Methods: MENA expression levels in the clinical HR<superscript>+</superscript> breast cancer samples (n = 119) were estimated using immunohistochemistry (IHC) to determine its association with the clinicopathological features, tamoxifen resistance, and survival outcomes. Western blotting (WB) and quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) analysis was performed to estimate the MENA protein and mRNA levels in the tamoxifen-sensitive and -resistant HR<superscript>+</superscript> breast cancer cell lines. Furthermore, CCK8, colony formation, and the transwell invasion and migration assays were used to analyze the effects of MENA knockdown on the biological behavior and tamoxifen sensitivity of the HR<superscript>+</superscript> breast cancer cell lines. Xenograft tumor experiments were performed in the nude mice to determine the tumor growth rates and tamoxifen sensitivity of the control and MENA knockdown HR<superscript>+</superscript> breast cancer cells in the presence and absence of tamoxifen treatment. Furthermore, we estimated the growth rates of organoids derived from the HR<superscript>+</superscript> breast cancer patients (n = 10) with high and low MENA expression levels when treated with tamoxifen. Results: HR<superscript>+</superscript> breast cancer patients with low MENA expression demonstrated tamoxifen resistance and poorer prognosis compared to those with high MENA expression. Univariate and multivariate Cox regression analysis demonstrated that MENA expression was an independent predictor of tamoxifen resistance in patients with HR<superscript>+</superscript> breast cancer. MENA knockdown HR<superscript>+</superscript> breast cancer cells showed significantly reduced tamoxifen sensitivity in the in vitro experiments and the in vivo xenograft tumor mouse model compared with the corresponding controls. Furthermore, MENA knockdown increased the in vitro invasion and migration of the HR<superscript>+</superscript> breast cancer cells. HR<superscript>+</superscript> breast cancer organoids with low MENA expression demonstrated reduced tamoxifen sensitivity than those with higher MENA expression. Mechanistically, P-AKT levels were significantly upregulated in the MENA-knockdown HR + breast cancer cells treated with or without 4-OHT compared with the corresponding controls. Conclusions: This study demonstrated that downregulation of MENA promoted tamoxifen resistance in the HR<superscript>+</superscript> breast cancer tissues and cells by enhancing the AKT signaling pathway. Therefore, MENA is a promising prediction biomarker for determining tamoxifen sensitivity in patients with HR<superscript>+</superscript> breast cancer. [ABSTRACT FROM AUTHOR]

Details

Language :
English
ISSN :
17456150
Volume :
19
Issue :
1
Database :
Complementary Index
Journal :
Biology Direct
Publication Type :
Academic Journal
Accession number :
175931117
Full Text :
https://doi.org/10.1186/s13062-024-00464-3